HAVCR2/TIM-3 belongs to the TIM family of immune regulators and is expressed on T cells (CD4⁺ Th1, CD8⁺), dendritic cells, macrophages, and NK cells . Its structure includes:
Extracellular domain: Immunoglobulin-like domain (IgV) for ligand binding (e.g., galectin-9, phosphatidylserine).
Mucin domain: Glycosylated region near the transmembrane domain.
Intracellular tail: Tyrosine residues critical for signaling and immune suppression .
Immune inhibition: Suppresses Th1 responses, promotes tolerance, and induces T-cell exhaustion .
Apoptotic cell clearance: Binds phosphatidylserine on dying cells to facilitate phagocytosis .
Cytokine regulation: Modulates IFN-γ, TNF-α, and IL-1β production .
Mouse models with targeted HAVCR2 mutations or knockouts have been developed to study its role in immunity and disease.
Note: The p.Y82C mutation (c.245A>G) is a germline variant linked to human SPTCL and HLH .
Enhanced Th1 responses: HAVCR2 KO mice show exacerbated experimental autoimmune encephalomyelitis (EAE) due to unregulated IFN-γ production .
Immune exhaustion: In chronic viral infections (e.g., LCMV), HAVCR2/PD-1 dual blockade restores CD8⁺ T-cell function .
Tumor microenvironment: HAVCR2 upregulation in tumor-infiltrating lymphocytes (TILs) correlates with immunotherapy resistance .
Combination therapies: Anti-HAVCR2 antibodies (e.g., TSR-022) are tested with anti-PD-1 in clinical trials to overcome resistance .
Cytokine storm: HAVCR2-mutant mice exhibit elevated IL-1β, TNF-α, and IFN-γ, driving HLH-like pathology .
Therapeutic response: JAK inhibitors (e.g., ruxolitinib) reduce cytokine levels and improve survival in HLH models .
Genetic predisposition: Biallelic HAVCR2 mutations (homozygous p.Y82C) are enriched in SPTCL patients, leading to lymphoproliferation and HLH .
Mechanism: Impaired TIM-3 expression disrupts Treg function and inflammasome regulation .
Genetic association: GWAS links HAVCR2 variants to late-onset Alzheimer’s, potentially via interactions with amyloid-β .
Hepatitis A virus cellular receptor 2, HAVcr-2, T-cell immunoglobulin and mucin domain-containing protein 3, TIMD-3, T-cell membrane protein 3, TIM-3, HAVCR2, TIM3, TIMD3, TIM3, KIM-3
HEK293 cells.
RSLENAYVFE VGKNAYLPCS YTLSTPGALV PMCWGKGFCP WSQCTNELLR TDERNVTYQK SSRYQLKGDL NKGDVSLIIK NVTLDDHGTY CCRIQFPGLM NDKKLELKLD IKAAKVTPAQ TAHGDSTTAS PRTLTTERNG SETQTLVTLH NNNGTKISTW ADEIKDSGET IRTALEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGKHHH HHH
Mouse HAVCR2/TIM-3 is a member of the immunoglobulin superfamily and TIM family of proteins. The protein contains an extracellular domain spanning from Leu22 to Arg191. The full recombinant protein has a predicted molecular weight of 19.9 kDa, but due to significant glycosylation, it typically migrates to 40-68 kDa when analyzed by Bis-Tris PAGE . The protein structure includes immunoglobulin and mucin domains that are critical for its binding interactions and downstream signaling functions .
While both mouse and human HAVCR2/TIM-3 belong to the same protein family and share functional similarities, there are important structural differences to consider when designing cross-species experiments. Both contain immunoglobulin and mucin domains, but amino acid sequence variations exist that may affect binding affinities to ligands. This is particularly important when testing therapeutic antibodies or ligand interactions that may not translate directly between species .
Mouse HAVCR2 is commonly referenced using several alternative identifiers:
These alternative designations are important to know when conducting literature searches or database queries to ensure comprehensive coverage of relevant research.
Mouse HAVCR2/TIM-3 is predominantly expressed on several immune cell populations:
CD4+ Th1 cells but not Th2 cells
Exhausted CD8+ T cells, particularly in chronic infection and cancer contexts
Natural killer (NK) cells
Dendritic cells
Regulatory T cells (Treg)
The expression is typically induced following T cell activation and is considered a marker of T cell exhaustion when co-expressed with other checkpoint molecules like PD-1 .
HAVCR2/TIM-3 expression is dynamically regulated in response to various stimuli. In T cells, its expression increases following activation and during chronic antigen exposure, correlating with an exhausted phenotype. The regulation involves transcriptional mechanisms that can be influenced by cytokine signaling pathways, particularly those associated with Th1 differentiation. In dendritic cells, expression can be modulated by exposure to danger-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) .
According to the International Mouse Phenotyping Consortium (IMPC) data, expression of HAVCR2 has been examined in 73 adult tissues, though specific lacZ expression data appears to be limited . The protein is predominantly expressed in immune tissues and cells rather than showing broad tissue distribution. Its expression in non-immune tissues under pathological conditions remains an active area of investigation.
Mouse HAVCR2/TIM-3 interacts with several ligands that mediate its diverse functions:
Galectin-9: A primary binding partner that induces immunosuppressive functions. The binding enhances immune tolerance and inhibits anti-tumor immunity. The ED50 for binding to human Galectin-9 in functional ELISA is less than 20 μg/ml .
Phosphatidylserine (PtSer): HAVCR2/TIM-3 can recognize phosphatidylserine on apoptotic cells in a calcium-dependent manner, facilitating their phagocytosis, particularly by dendritic cells .
CEACAM-1: Heterodimerization with CEACAM-1 appears critical for the inhibitory function of TIM-3. Co-blockade of TIM-3 and CEACAM-1 has shown enhanced antitumor responses in mouse models .
HMGB1: In tumor-infiltrating dendritic cells, TIM-3 can interact with HMGB1, interfering with nucleic acid sensing and trafficking to endosomes .
HAVCR2/TIM-3 signaling in mouse T cells has complex effects:
Inhibitory Functions: Generally accepted to have an inhibitory role, TIM-3 attenuates TCR-induced signaling, specifically by blocking NF-κB and NFAT promoter activities, resulting in reduced IL-2 secretion. This function may involve its association with LCK, which impairs phosphorylation of TCR subunits .
T Cell Exhaustion: When co-expressed with PD-1 on tumor-infiltrating T cells, TIM-3 contributes to a severe exhausted phenotype, characterized by reduced proliferation and cytokine production .
Apoptosis Induction: Binding to Galectin-9 can induce apoptosis of antigen-specific T cells, potentially involving TIM-3 phosphorylation and disruption of its association with BAG6 .
Context-Dependent Activation: In contrast to its inhibitory functions, TIM-3 has also been shown to activate TCR-induced signaling in some contexts, potentially involving ZAP70, LCP2, LCK, and FYN .
In mouse dendritic cells (DCs), HAVCR2/TIM-3 has several important functions:
Phagocytosis of Apoptotic Cells: TIM-3 mediates the engulfment of apoptotic cells by DCs through recognition of phosphatidylserine .
Antigen Cross-Presentation: TIM-3 facilitates the cross-presentation of antigens from apoptotic cells .
Immunoregulation: TIM-3 expressed on DCs can positively regulate innate immune responses and, in synergy with Toll-like receptors, promote TNF-α secretion .
Suppression of Nucleic Acid Sensing: In tumor-infiltrating DCs, TIM-3 can suppress nucleic acid-mediated innate immune responses by interacting with HMGB1 and interfering with nucleic acid sensing and trafficking to endosomes .
For optimal handling of recombinant mouse HAVCR2/TIM-3 protein:
Reconstitution:
Storage Conditions:
Store at -20°C to -80°C for 12 months as supplied from date of receipt
Storage at -80°C for 3 months after reconstitution
Aliquot the protein into smaller quantities for optimal storage
Minimize freeze-thaw cycles as repeated freezing and thawing is not recommended
Several methodological approaches are effective for studying mouse HAVCR2/TIM-3:
Genetic Models:
Protein Interaction Studies:
Functional Assays:
In vivo Models:
Researchers often encounter several challenges when studying mouse HAVCR2/TIM-3:
Protein Glycosylation Variability:
Context-Dependent Functions:
Ligand Specificity:
Species Differences:
In mouse cancer models, HAVCR2/TIM-3 plays several important roles:
T Cell Exhaustion: TIM-3 is expressed by exhausted T cells in cancer settings. Tumor-infiltrating T cells that co-express PD-1 and TIM-3 exhibit the most severe exhausted phenotype, characterized by diminished anti-tumor activity .
Dendritic Cell Dysfunction: TIM-3 expression on tumor-infiltrating dendritic cells suppresses innate immunity by reducing the immunogenicity of nucleic acids released by dying tumor cells, thereby contributing to immune evasion .
Regulatory T Cell Function: TIM-3 expressed on Treg cells can inhibit effector T cell responses, including Th17 responses, further dampening anti-tumor immunity .
Therapeutic Target: Blockade of TIM-3, particularly in combination with other checkpoint inhibitors, has shown promise in enhancing anti-tumor responses in various mouse cancer models .
HAVCR2/TIM-3 has significant implications in chronic infection models:
T Cell Exhaustion Marker: Similar to cancer contexts, TIM-3 is upregulated on exhausted T cells during chronic viral infections, contributing to impaired clearance of persistent pathogens .
Antimicrobial Responses: TIM-3 expressed on Th1 cells interacts with Galectin-9 on Mycobacterium tuberculosis-infected macrophages to stimulate antibactericidal activity, including IL-1β secretion, and to restrict intracellular bacterial growth .
Acute Infection Responses: TIM-3 has been reported to enhance CD8+ T cell responses to acute infections such as Listeria monocytogenes, highlighting its context-dependent functionality .
NK Cell Regulation: TIM-3 expressed on NK cells can enhance IFN-γ production in response to certain pathogens but can also suppress NK cell-mediated cytotoxicity in other contexts, demonstrating its complex role in innate immunity .
According to the International Mouse Phenotyping Consortium (IMPC), HAVCR2 knockout mice exhibit specific phenotypes:
Significant Phenotypes: Two significant phenotypes have been identified in HAVCR2 knockout mice, though detailed descriptions are not provided in the available search results .
Associated Diseases: Two diseases are associated with HAVCR2 mutations, based on comparisons of mouse phenotypes with human disease phenotypes .
Physiological Systems Impact: The knockout appears to significantly impact certain physiological systems, though specific details would require further investigation .
Autoimmunity Predisposition: Based on the known functions of TIM-3 in regulating T cell responses, knockout mice may exhibit enhanced susceptibility to autoimmune conditions due to dysregulated Th1 responses .
The interaction between HAVCR2/TIM-3 and other molecules, particularly CEACAM-1, presents interesting therapeutic opportunities:
Co-targeting Strategies: Research indicates that heterodimerization of TIM-3 with CEACAM-1 is critical for the inhibitory function of TIM-3. Co-blockade of TIM-3 and CEACAM-1 has enhanced antitumor responses in a mouse model of colorectal cancer .
Structure-Based Drug Design: Understanding the structural basis of these heterodimeric interactions could inform the development of small molecules or biologics that specifically disrupt these interactions while preserving beneficial TIM-3 functions.
Combination Therapies: Exploring combinations of TIM-3 blockade with other checkpoint inhibitors (beyond PD-1) based on heterodimeric interactions could identify synergistic therapeutic approaches for cancer immunotherapy.
Cell Type-Specific Targeting: Developing strategies to target TIM-3 heterodimeric complexes on specific cell populations (e.g., tumor-infiltrating dendritic cells vs. T cells) might improve therapeutic specificity and reduce off-target effects.
The capacity of HAVCR2/TIM-3 to bind phosphatidylserine (PtSer) on apoptotic cells has significant implications for efferocytosis research:
Mechanistic Studies: Investigating the calcium-dependent binding mechanism of TIM-3 to PtSer could illuminate general principles of phospholipid recognition by immune receptors.
Cell Type-Specific Effects: While TIM-3 on dendritic cells mediates engulfment of apoptotic cells, TIM-3 on T cells promotes conjugation but not engulfment. Understanding this differential response could reveal important cell-specific signaling mechanisms .
Cross-Presentation Pathways: Exploring how TIM-3-mediated uptake of apoptotic cells influences antigen processing and cross-presentation pathways in dendritic cells could inform vaccine design and cancer immunotherapy approaches.
Resolution of Inflammation: Investigating the role of TIM-3 in efferocytosis during the resolution phase of inflammatory responses could identify new approaches for treating inflammatory diseases.
Single-cell technologies offer powerful approaches to address current knowledge gaps in HAVCR2/TIM-3 biology:
Heterogeneity in Expression Patterns: Single-cell RNA sequencing could reveal previously unrecognized heterogeneity in TIM-3 expression across immune cell populations and within conventionally defined subsets.
Temporal Dynamics: Single-cell trajectory analysis could elucidate the temporal dynamics of TIM-3 expression during T cell activation, exhaustion, and memory formation.
Spatial Context: Spatial transcriptomics approaches could map TIM-3 expression in relation to other immune cells within tissues, particularly in tumor microenvironments and sites of chronic infection.
Protein-Protein Interactions: Single-cell proteomics and proximity labeling approaches could identify cell type-specific protein interaction networks for TIM-3, potentially revealing new binding partners and signaling mechanisms.
When designing experiments involving mouse HAVCR2/TIM-3, researchers should consider the following critical controls:
Specificity Controls: Include isotype controls for antibodies and use HAVCR2 knockout cells or tissues to confirm signal specificity.
Glycosylation Considerations: Given the extensive glycosylation of TIM-3, researchers should consider how glycosylation status might affect experimental outcomes, particularly in binding studies.
Context Documentation: Clearly document the cellular context and activation state, as TIM-3 functions can vary significantly depending on the cell type and environmental conditions.
Ligand Validation: When studying ligand interactions, validate binding using multiple methodologies given controversies surrounding some reported TIM-3 ligands.
Cross-Species Validation: For translational studies, validate key findings across mouse and human systems to account for species-specific differences in TIM-3 biology.
Several emerging technologies hold promise for advancing our understanding of mouse HAVCR2/TIM-3:
CRISPR-Based Screening: Genome-wide CRISPR screens in primary mouse immune cells could identify novel regulators of TIM-3 expression and function.
Intravital Imaging: Advanced intravital microscopy techniques could track TIM-3-expressing cells in vivo during immune responses, providing insights into dynamic cellular interactions.
Protein Engineering: Engineered variants of TIM-3 with altered binding properties could help dissect the relative contributions of different ligand interactions to TIM-3 function.
Organoid Systems: Immune-competent organoid systems could provide more physiologically relevant platforms for studying TIM-3 in tissue-specific contexts.
Systems Biology Approaches: Integrative computational analyses combining transcriptomic, proteomic, and functional data could reveal emergent properties of TIM-3 signaling networks not apparent from reductionist approaches.
The continued investigation of HAVCR2/TIM-3 in mouse models remains essential for understanding fundamental immunological processes and developing novel therapeutic approaches for cancer, chronic infections, and autoimmune diseases.
The HAVCR2 gene encodes a type I membrane protein that consists of an immunoglobulin variable-like (IgV) domain, a mucin-like domain, and a cytoplasmic domain with a tyrosine phosphorylation motif . The protein is approximately 301 amino acids in length and shares significant homology with the hepatitis A virus cellular receptor 1 (HAVCR1), also known as TIM-1 or Kim1 .
HAVCR2 is a critical negative regulator in the immune system, acting as a checkpoint in peripheral tolerance and innate immune and inflammatory responses . It is specifically expressed on activated Th1 cells and CD11b+ macrophages . The protein regulates macrophage activation and enhances the severity of experimental autoimmune encephalomyelitis (EAE) in mice .
HAVCR2 is related to the hepatitis A virus cellular receptor (HAVCR1), which the hepatitis A virus (HAV) uses to infect cells . The interaction of HAV with soluble forms of its cellular receptor shares the physiological requirements of infectivity in cell culture . This interaction is crucial for understanding the cell entry process of HAV and its pathogenesis .
Recombinant forms of HAVCR2, such as the mouse recombinant version, are used in research to study its function and interaction with various ligands. These recombinant proteins are valuable tools for investigating the molecular mechanisms underlying immune responses and viral infections.
The role of HAVCR2 in regulating immune responses makes it a potential target for therapeutic interventions in autoimmune diseases and cancer. By modulating the activity of HAVCR2, it may be possible to enhance immune responses against tumors or reduce inflammation in autoimmune conditions.